Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
Add more filters

Publication year range
1.
Cell ; 139(7): 1243-54, 2009 Dec 24.
Article in English | MEDLINE | ID: mdl-20064371

ABSTRACT

Influenza viruses exploit host cell machinery to replicate, resulting in epidemics of respiratory illness. In turn, the host expresses antiviral restriction factors to defend against infection. To find host cell modifiers of influenza A H1N1 viral infection, we used a functional genomic screen and identified over 120 influenza A virus-dependency factors with roles in endosomal acidification, vesicular trafficking, mitochondrial metabolism, and RNA splicing. We discovered that the interferon-inducible transmembrane proteins IFITM1, 2, and 3 restrict an early step in influenza A viral replication. The IFITM proteins confer basal resistance to influenza A virus but are also inducible by interferons type I and II and are critical for interferon's virustatic actions. Further characterization revealed that the IFITM proteins inhibit the early replication of flaviviruses, including dengue virus and West Nile virus. Collectively this work identifies a family of antiviral restriction factors that mediate cellular innate immunity to at least three major human pathogens.


Subject(s)
Flavivirus Infections/immunology , Influenza, Human/immunology , Membrane Proteins/immunology , Animals , Antigens, Differentiation , Cell Line, Tumor , Dengue Virus/immunology , Humans , Immunity, Innate , Influenza A virus/immunology , Interferons/immunology , Mice , RNA-Binding Proteins/immunology , West Nile virus/immunology , West Nile virus/physiology
2.
Nature ; 541(7636): 233-236, 2017 01 12.
Article in English | MEDLINE | ID: mdl-28052056

ABSTRACT

Metastasis is the leading cause of death for cancer patients. This multi-stage process requires tumour cells to survive in the circulation, extravasate at distant sites, then proliferate; it involves contributions from both the tumour cell and tumour microenvironment ('host', which includes stromal cells and the immune system). Studies suggest the early steps of the metastatic process are relatively efficient, with the post-extravasation regulation of tumour growth ('colonization') being critical in determining metastatic outcome. Here we show the results of screening 810 mutant mouse lines using an in vivo assay to identify microenvironmental regulators of metastatic colonization. We identify 23 genes that, when disrupted in mouse, modify the ability of tumour cells to establish metastatic foci, with 19 of these genes not previously demonstrated to play a role in host control of metastasis. The largest reduction in pulmonary metastasis was observed in sphingosine-1-phosphate (S1P) transporter spinster homologue 2 (Spns2)-deficient mice. We demonstrate a novel outcome of S1P-mediated regulation of lymphocyte trafficking, whereby deletion of Spns2, either globally or in a lymphatic endothelial-specific manner, creates a circulating lymphopenia and a higher percentage of effector T cells and natural killer (NK) cells present in the lung. This allows for potent tumour cell killing, and an overall decreased metastatic burden.


Subject(s)
Anion Transport Proteins/genetics , Anion Transport Proteins/metabolism , Genome/genetics , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Animals , Anion Transport Proteins/deficiency , Cell Line, Tumor , Cell Movement , Disease Models, Animal , Female , Genomics , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lymphopenia/genetics , Lymphopenia/pathology , Lysophospholipids/metabolism , Male , Mice , Sphingosine/analogs & derivatives , Sphingosine/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Tumor Microenvironment
3.
Blood ; 136(20): 2319-2333, 2020 11 12.
Article in English | MEDLINE | ID: mdl-32573712

ABSTRACT

Karyotype is an important prognostic factor in childhood B-cell precursor acute lymphoblastic leukemia (BCP-ALL), but the underlying pharmacogenomics remain unknown. Asparaginase is an integral component in current chemotherapy for childhood BCP-ALL. Asparaginase therapy depletes serum asparagine. Normal hematopoietic cells can produce asparagine by asparagine synthetase (ASNS) activity, but ALL cells are unable to synthesize adequate amounts of asparagine. The ASNS gene has a typical CpG island in its promoter. Thus, methylation of the ASNS CpG island could be one of the epigenetic mechanisms for ASNS gene silencing in BCP-ALL. To gain deep insights into the pharmacogenomics of asparaginase therapy, we investigated the association of ASNS methylation status with asparaginase sensitivity. The ASNS CpG island is largely unmethylated in normal hematopoietic cells, but it is allele-specifically methylated in BCP-ALL cells. The ASNS gene is located at 7q21, an evolutionally conserved imprinted gene cluster. ASNS methylation in childhood BCP-ALL is associated with an aberrant methylation of the imprinted gene cluster at 7q21. Aberrant methylation of mouse Asns and a syntenic imprinted gene cluster is also confirmed in leukemic spleen samples from ETV6-RUNX1 knockin mice. In 3 childhood BCP-ALL cohorts, ASNS is highly methylated in BCP-ALL patients with favorable karyotypes but is mostly unmethylated in BCP-ALL patients with poor prognostic karyotypes. Higher ASNS methylation is associated with higher L-asparaginase sensitivity in BCP-ALL through lower ASNS gene and protein expression levels. These observations demonstrate that silencing of the ASNS gene as a result of aberrant imprinting is a pharmacogenetic mechanism for the leukemia-specific activity of asparaginase therapy in BCP-ALL.


Subject(s)
Asparaginase/therapeutic use , Aspartate-Ammonia Ligase/genetics , Pharmacogenomic Variants/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Animals , Child , Chromosome Aberrations , DNA Methylation/genetics , Genomic Imprinting/genetics , Humans , Mice
4.
Histopathology ; 80(1): 135-149, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34958502

ABSTRACT

Primary cutaneous and mucosal melanoma shows a wide histological spectrum. The correct diagnosis depends upon the demonstration of melanocytic differentiation by recognition of an associated in-situ component or immunohistochemical evidence of a melanocytic phenotype using conventional melanocytic markers, such as S-100, SOX10, Melan-A and HMB-45. Exceptionally, melanomas lose their melanocytic phenotype, at least focally, and show differentiation towards other lineages. Review of the literature shows that de- and trans-differentiation in melanoma is rare but probably under-recognised and under-reported. These often large and frequently ulcerated tumours affect adults and show a wide anatomical distribution, including mucosal sites, although there is a predilection for sun-damaged skin of the head and neck. Histologically, the tumours are biphasic and contain a pre-existing conventional melanoma. The de-differentiated component closely resembles atypical fibroxanthoma, both morphologically and immunohistochemically. Trans-differentiated melanoma may show rhabdomyosarcomatous or spindle cell carcinomatous features. Undifferentiated melanomas are similar tumours in which the conventional melanoma component is absent. Their diagnosis depends entirely upon the clinical context and identification of a classical melanoma driver gene mutation, i.e. BRAF V600E. The diagnosis of these rare and unusual tumours is challenging, and requires thorough tumour sampling and recognition of the background of a pre-existing but often focal conventional melanoma together with molecular analysis.


Subject(s)
Cell Dedifferentiation/physiology , Cell Differentiation/physiology , Melanoma/pathology , Skin Neoplasms/pathology , Skin/pathology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Humans , Melanoma/genetics , Melanoma/metabolism , Mutation , Skin/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/metabolism
5.
Mod Pathol ; 34(11): 2009-2019, 2021 11.
Article in English | MEDLINE | ID: mdl-34155350

ABSTRACT

Dedifferentiation and transdifferentiation are rare and only poorly understood phenomena in cutaneous melanoma. To study this disease more comprehensively we have retrieved 11 primary cutaneous melanomas from our pathology archives showing biphasic features characterized by a conventional melanoma and additional areas of de-/trans-differentiation as defined by a lack of immunohistochemical expression of all conventional melanocytic markers (S-100 protein, SOX10, Melan-A, and HMB-45). The clinical, histologic, and immunohistochemical findings were recorded and follow-up was obtained. The patients were mostly elderly (median: 81 years; range: 42-86 years) without significant gender predilection, and the sun-exposed skin of the head and neck area was most commonly affected. The tumors were deeply invasive with a mean depth of 7 mm (range: 4-80 mm). The dedifferentiated component showed atypical fibroxanthoma-like features in the majority of cases (7), while additional rhabdomyosarcomatous and epithelial transdifferentiation was noted histologically and/or immunohistochemically in two tumors each. The background conventional melanoma component was of desmoplastic (4), superficial spreading (3), nodular (2), lentigo maligna (1), or spindle cell (1) types. For the seven patients with available follow-up data (median follow-up period of 25 months; range: 8-36 months), two died from their disease, and three developed metastases. Next-generation sequencing of the cohort revealed somatic mutations of established melanoma drivers including mainly NF1 mutations (5) in the conventional component, which was also detected in the corresponding de-/trans-differentiated component. In summary, the diagnosis of primary cutaneous de-/trans-differentiated melanoma is challenging and depends on the morphologic identification of conventional melanoma. Molecular analysis is diagnostically helpful as the mutated gene profile is shared between the conventional and de-/trans-differentiated components. Importantly, de-/trans-differentiation does not appear to confer a more aggressive behavior.


Subject(s)
Genomics , Melanoma/pathology , Neurofibromin 1/genetics , Skin Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Cell Differentiation , DNA, Neoplasm/genetics , Diagnosis, Differential , Female , High-Throughput Nucleotide Sequencing , Humans , Male , Melanoma/genetics , Melanoma/metabolism , Middle Aged , Neoplasm Proteins/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/metabolism
6.
J Pathol ; 252(1): 4-21, 2020 09.
Article in English | MEDLINE | ID: mdl-32652526

ABSTRACT

In contrast to other cancer types, melanoma incidence has been increasing over the last 50 years, and while it still represents less than 5% of all cutaneous malignancies, melanoma accounts for the majority of skin cancer deaths, due to its propensity to metastasise. Whilst melanoma most commonly affects the skin, it can also arise in mucosal surfaces, the eye, and the brain. For new therapies to be developed, a better understanding of the genetic landscape, signalling pathways, and tumour-microenvironmental interactions is needed. This is where animal models are of critical importance. The mouse is the foremost used model of human melanoma. Arguably this is due to its plethora of benefits as a laboratory animal; however, it is important to note that unlike humans, melanocytes are not present at the dermal-epidermal junction in mice and mice do not develop melanoma without genetic manipulation. In contrast, there are numerous reports of animals that spontaneously develop melanoma, ranging from sharks and parrots to hippos and monkeys. In addition, several domesticated and laboratory-bred animals spontaneously develop melanoma or UV-induced melanoma, specifically, fish, opossums, pigs, horses, cats, and dogs. In this review, we look at spontaneously occurring animal 'models' of melanoma and discuss their relevance to the different types of melanoma found in humans. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland..


Subject(s)
Cat Diseases/pathology , Dog Diseases/pathology , Fish Diseases/pathology , Horse Diseases/pathology , Melanoma/veterinary , Skin Neoplasms/veterinary , Swine Diseases/pathology , Animals , Cat Diseases/genetics , Cats , Dog Diseases/genetics , Dogs , Fish Diseases/genetics , Horse Diseases/genetics , Horses , Humans , Melanoma/genetics , Melanoma/pathology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Swine , Swine Diseases/genetics
7.
PLoS Pathog ; 14(4): e1006968, 2018 04.
Article in English | MEDLINE | ID: mdl-29698475

ABSTRACT

Approximately 12% of all human cancers worldwide are caused by infections with oncogenic viruses. Kaposi's sarcoma herpesvirus/human herpesvirus 8 (KSHV/HHV8) is one of the oncogenic viruses responsible for human cancers, including Kaposi's sarcoma (KS), Primary Effusion Lymphoma (PEL), and the lymphoproliferative disorder multicentric Castleman's disease (MCD). Chronic inflammation mediated by KSHV infection plays a decisive role in the development and survival of these cancers. NF-κB, a family of transcription factors regulating inflammation, cell survival, and proliferation, is persistently activated in KSHV-infected cells. The KSHV latent and lytic expressing oncogenes involved in NF-κB activation are vFLIP/K13 and vGPCR, respectively. However, the mechanisms by which NF-κB is activated by vFLIP and vGPCR are poorly understood. In this study, we have found that a host molecule, Cell Adhesion Molecule 1 (CADM1), is robustly upregulated in KSHV-infected PBMCs and KSHV-associated PEL cells. Further investigation determined that both vFLIP and vGPCR interacted with CADM1. The PDZ binding motif localized at the carboxyl terminus of CADM1 is essential for both vGPCR and vFLIP to maintain chronic NF-κB activation. Membrane lipid raft associated CADM1 interaction with vFLIP is critical for the initiation of IKK kinase complex and NF-κB activation in the PEL cells. In addition, CADM1 played essential roles in the survival of KSHV-associated PEL cells. These data indicate that CADM1 plays key roles in the activation of NF-κB pathways during latent and lytic phases of the KSHV life cycle and the survival of KSHV-infected cells.


Subject(s)
Cell Adhesion Molecule-1/metabolism , Lymphoma, Primary Effusion/metabolism , NF-kappa B/metabolism , Receptors, Chemokine/metabolism , Sarcoma, Kaposi/metabolism , Viral Proteins/metabolism , Cell Adhesion Molecule-1/genetics , Herpesvirus 8, Human/pathogenicity , Humans , Lymphoma, Primary Effusion/genetics , Lymphoma, Primary Effusion/virology , NF-kappa B/genetics , Receptors, Chemokine/genetics , Sarcoma, Kaposi/genetics , Sarcoma, Kaposi/virology , Tumor Cells, Cultured , Viral Proteins/genetics
8.
J Pathol ; 246(3): 300-310, 2018 11.
Article in English | MEDLINE | ID: mdl-30062795

ABSTRACT

Metastasis is the leading cause of death in cancer patients, and successful colonisation of a secondary organ by circulating tumour cells (CTCs) is the rate-limiting step of this process. We used tail-vein injection of B16-F10 melanoma cells into mice to mimic the presence of CTCs and to allow for the assessment of host (microenvironmental) factors that regulate pulmonary metastatic colonisation. We found that mice deficient for the individual subunits of the NADPH oxidase of myeloid cells, NOX2 (encoded by Cyba, Cybb, Ncf1, Ncf2, and Ncf4), all showed decreased pulmonary metastatic colonisation. To understand the role of NOX2 in controlling tumour cell survival in the pulmonary microenvironment, we focused on Cyba-deficient (Cybatm1a ) mice, which showed the most significant decrease in metastatic colonisation. Interestingly, histological assessment of pulmonary metastatic colonisation was not possible in Cybatm1a mice, owing to the presence of large granulomas composed of galectin-3 (Mac-2)-positive macrophages and eosinophilic deposits; granulomas of variable penetrance and severity were also found in Cybatm1a mice that were not injected with melanoma cells, and these contributed to their decreased survival. The decreased pulmonary metastatic colonisation of Cybatm1a mice was not due to any overt defects in vascular permeability, and bone marrow chimaeras confirmed a role for the haematological system in the reduced metastatic colonisation phenotype. Examination of the lymphocyte populations, which are known key regulators of metastatic colonisation, revealed an enhanced proportion of activated T and natural killer cells in the lungs of Cybatm1a mice, relative to controls. The reduced metastatic colonisation, presence of granulomas and altered immune cell populations observed in Cybatm1a lungs were mirrored in Ncf2-deficient (Ncf2tm1a ) mice. Thus, we show that NOX2 deficiency results in both granulomas and the accumulation of antitumoural immune cells in the lungs that probably mediate the decreased pulmonary metastatic colonisation. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Cell Movement , Cytochrome b Group/deficiency , Granuloma/pathology , Lung Neoplasms/secondary , Melanoma, Experimental/secondary , NADPH Oxidase 2/deficiency , NADPH Oxidases/deficiency , Neoplastic Cells, Circulating/pathology , Animals , Cell Line, Tumor , Cytochrome b Group/genetics , Granuloma/enzymology , Granuloma/genetics , Granuloma/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Melanoma, Experimental/enzymology , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Mice, Knockout , NADPH Oxidase 2/genetics , NADPH Oxidases/genetics , Neoplasm Invasiveness , Neoplastic Cells, Circulating/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Tumor Microenvironment
9.
Proc Natl Acad Sci U S A ; 113(29): 8290-5, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27357679

ABSTRACT

Prostate cancer (CaP) is the most common adult male cancer in the developed world. The paucity of biomarkers to predict prostate tumor biology makes it important to identify key pathways that confer poor prognosis and guide potential targeted therapy. Using a murine forward mutagenesis screen in a Pten-null background, we identified peroxisome proliferator-activated receptor gamma (Pparg), encoding a ligand-activated transcription factor, as a promoter of metastatic CaP through activation of lipid signaling pathways, including up-regulation of lipid synthesis enzymes [fatty acid synthase (FASN), acetyl-CoA carboxylase (ACC), ATP citrate lyase (ACLY)]. Importantly, inhibition of PPARG suppressed tumor growth in vivo, with down-regulation of the lipid synthesis program. We show that elevated levels of PPARG strongly correlate with elevation of FASN in human CaP and that high levels of PPARG/FASN and PI3K/pAKT pathway activation confer a poor prognosis. These data suggest that CaP patients could be stratified in terms of PPARG/FASN and PTEN levels to identify patients with aggressive CaP who may respond favorably to PPARG/FASN inhibition.


Subject(s)
Fatty Acid Synthase, Type I/metabolism , PPAR gamma/metabolism , PTEN Phosphohydrolase/metabolism , Prostatic Neoplasms/metabolism , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Humans , Lipid Metabolism , Male , Mice , Middle Aged , PPAR gamma/genetics , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Transposases
10.
Nature ; 486(7402): 266-70, 2012 Apr 29.
Article in English | MEDLINE | ID: mdl-22699621

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) remains a lethal malignancy despite much progress concerning its molecular characterization. PDA tumours harbour four signature somatic mutations in addition to numerous lower frequency genetic events of uncertain significance. Here we use Sleeping Beauty (SB) transposon-mediated insertional mutagenesis in a mouse model of pancreatic ductal preneoplasia to identify genes that cooperate with oncogenic Kras(G12D) to accelerate tumorigenesis and promote progression. Our screen revealed new candidate genes for PDA and confirmed the importance of many genes and pathways previously implicated in human PDA. The most commonly mutated gene was the X-linked deubiquitinase Usp9x, which was inactivated in over 50% of the tumours. Although previous work had attributed a pro-survival role to USP9X in human neoplasia, we found instead that loss of Usp9x enhances transformation and protects pancreatic cancer cells from anoikis. Clinically, low USP9X protein and messenger RNA expression in PDA correlates with poor survival after surgery, and USP9X levels are inversely associated with metastatic burden in advanced disease. Furthermore, chromatin modulation with trichostatin A or 5-aza-2'-deoxycytidine elevates USP9X expression in human PDA cell lines, indicating a clinical approach for certain patients. The conditional deletion of Usp9x cooperated with Kras(G12D) to accelerate pancreatic tumorigenesis in mice, validating their genetic interaction. We propose that USP9X is a major tumour suppressor gene with prognostic and therapeutic relevance in PDA.


Subject(s)
Carcinoma, Pancreatic Ductal/enzymology , Pancreatic Neoplasms/enzymology , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Animals , Anoikis/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Disease Models, Animal , Endopeptidases , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred C57BL , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , U937 Cells
11.
Genes Dev ; 24(22): 2505-16, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20971813

ABSTRACT

During female meiosis, bivalent chromosomes are thought to be held together from birth until ovulation by sister chromatid cohesion mediated by cohesin complexes whose ring structure depends on kleisin subunits, either Rec8 or Scc1. Because cohesion is established at DNA replication in the embryo, its maintenance for such a long time may require cohesin turnover. To address whether Rec8- or Scc1-containing cohesin holds bivalents together and whether it turns over, we created mice whose kleisin subunits can be cleaved by TEV protease. We show by microinjection experiments and confocal live-cell imaging that Rec8 cleavage triggers chiasmata resolution during meiosis I and sister centromere disjunction during meiosis II, while Scc1 cleavage triggers sister chromatid disjunction in the first embryonic mitosis, demonstrating a dramatic transition from Rec8- to Scc1-containing cohesin at fertilization. Crucially, activation of an ectopic Rec8 transgene during the growing phase of Rec8(TEV)(/TEV) oocytes does not prevent TEV-mediated bivalent destruction, implying little or no cohesin turnover for ≥2 wk during oocyte growth. We suggest that the inability of oocytes to regenerate cohesion may contribute to age-related meiosis I errors.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Chromosomes/metabolism , Nuclear Proteins/metabolism , Oocytes/growth & development , Oocytes/metabolism , Phosphoproteins/metabolism , Animals , Cells, Cultured , Centromere/genetics , Chromosomes/genetics , Endopeptidases/metabolism , Female , Mice , Nuclear Proteins/genetics , Phosphoproteins/genetics , Cohesins
12.
PLoS Pathog ; 11(3): e1004721, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25774694

ABSTRACT

Persistent activation of NF-κB by the Human T-cell leukemia virus type 1 (HTLV-1) oncoprotein, Tax, is vital for the development and pathogenesis of adult T-cell leukemia (ATL) and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). K63-linked polyubiquitinated Tax activates the IKK complex in the plasma membrane-associated lipid raft microdomain. Tax also interacts with TAX1BP1 to inactivate the NF-κB negative regulatory ubiquitin-editing A20 enzyme complex. However, the molecular mechanisms of Tax-mediated IKK activation and A20 protein complex inactivation are poorly understood. Here, we demonstrated that membrane associated CADM1 (Cell adhesion molecule1) recruits Ubc13 to Tax, causing K63-linked polyubiquitination of Tax, and IKK complex activation in the membrane lipid raft. The c-terminal cytoplasmic tail containing PDZ binding motif of CADM1 is critical for Tax to maintain persistent NF-κB activation. Finally, Tax failed to inactivate the NF-κB negative regulator ubiquitin-editing enzyme A20 complex, and activate the IKK complex in the lipid raft in absence of CADM1. Our results thus indicate that CADM1 functions as a critical scaffold molecule for Tax and Ubc13 to form a cellular complex with NEMO, TAX1BP1 and NRP, to activate the IKK complex in the plasma membrane-associated lipid rafts, to inactivate NF-κB negative regulators, and maintain persistent NF-κB activation in HTLV-1 infected cells.


Subject(s)
Cell Adhesion Molecules/metabolism , Deltaretrovirus Infections/metabolism , Genes, pX/physiology , Immunoglobulins/metabolism , NF-kappa B/metabolism , Signal Transduction/physiology , Animals , Cell Adhesion Molecule-1 , Cysteine Endopeptidases/metabolism , DNA-Binding Proteins/metabolism , Electrophoretic Mobility Shift Assay , Human T-lymphotropic virus 1 , Humans , Immunoblotting , Immunoprecipitation , Intracellular Signaling Peptides and Proteins/metabolism , Jurkat Cells , Mice , Mice, Knockout , Microscopy, Confocal , Nuclear Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Tumor Necrosis Factor alpha-Induced Protein 3 , Ubiquitin-Conjugating Enzymes/metabolism
13.
Blood ; 125(23): 3609-17, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-25855603

ABSTRACT

Alterations of genes encoding transcriptional regulators of lymphoid development are a hallmark of B-progenitor acute lymphoblastic leukemia (B-ALL) and most commonly involve PAX5, encoding the DNA-binding transcription factor paired-box 5. The majority of PAX5 alterations in ALL are heterozygous, and key PAX5 target genes are expressed in leukemic cells, suggesting that PAX5 may be a haploinsufficient tumor suppressor. To examine the role of PAX5 alterations in leukemogenesis, we performed mutagenesis screens of mice heterozygous for a loss-of-function Pax5 allele. Both chemical and retroviral mutagenesis resulted in a significantly increased penetrance and reduced latency of leukemia, with a shift to B-lymphoid lineage. Genomic profiling identified a high frequency of secondary genomic mutations, deletions, and retroviral insertions targeting B-lymphoid development, including Pax5, and additional genes and pathways mutated in ALL, including tumor suppressors, Ras, and Janus kinase-signal transducer and activator of transcription signaling. These results show that in contrast to simple Pax5 haploinsufficiency, multiple sequential alterations targeting lymphoid development are central to leukemogenesis and contribute to the arrest in lymphoid maturation characteristic of ALL. This cross-species analysis also validates the importance of concomitant alterations of multiple cellular growth, signaling, and tumor suppression pathways in the pathogenesis of B-ALL.


Subject(s)
Gene Deletion , Neoplasms, Experimental/metabolism , PAX5 Transcription Factor/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Mice , Mice, Mutant Strains , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , PAX5 Transcription Factor/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Tumor Suppressor Proteins/genetics
14.
J Pathol ; 238(2): 152-65, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26354726

ABSTRACT

Although transformation of melanocytes to melanoma is rare, the rapid growth, systemic spread, as well as the chemoresistance of melanoma present significant challenges for patient care. Here we review animal models of melanoma, including murine, canine, equine, and zebrafish models, and detail the immense contribution these models have made to our knowledge of human melanoma development, and to melanocyte biology. We also highlight the opportunities for cross-species comparative genomic studies of melanoma to identify the key molecular events that drive this complex disease.


Subject(s)
Disease Models, Animal , Melanoma/genetics , Skin Neoplasms/genetics , Animals , Cell Line, Tumor , Disease Progression , Dogs , Genetic Predisposition to Disease/genetics , Genome , Horses , Humans , Melanoma/therapy , Mice , Mice, Transgenic , Mutation/genetics , Neoplasm Transplantation , Skin Neoplasms/therapy , Transplantation, Heterologous , Uveal Neoplasms/genetics , Uveal Neoplasms/therapy , Zebrafish
15.
J Pathol ; 239(3): 374-83, 2016 07.
Article in English | MEDLINE | ID: mdl-27126290

ABSTRACT

The epidermis is the outermost layer of skin that acts as a barrier to protect the body from the external environment and to control water and heat loss. This barrier function is established through the multistage differentiation of keratinocytes and the presence of bioactive sphingolipids such as ceramides, the levels of which are tightly regulated by a balance of ceramide synthase and ceramidase activities. Here we reveal the essential role of alkaline ceramidase 1 (Acer1) in the skin. Acer1-deficient (Acer1(-/-) ) mice showed elevated levels of ceramide in the skin, aberrant hair shaft cuticle formation and cyclic alopecia. We demonstrate that Acer1 is specifically expressed in differentiated interfollicular epidermis, infundibulum and sebaceous glands and consequently Acer1(-/-) mice have significant alterations in infundibulum and sebaceous gland architecture. Acer1(-/-) skin also shows perturbed hair follicle stem cell compartments. These alterations result in Acer1(-/-) mice showing increased transepidermal water loss and a hypermetabolism phenotype with associated reduction of fat content with age. We conclude that Acer1 is indispensable for mammalian skin homeostasis and whole-body energy homeostasis. © 2016 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Alkaline Ceramidase/metabolism , Alopecia/enzymology , Ceramides/metabolism , Energy Metabolism , Homeostasis , Alkaline Ceramidase/genetics , Alopecia/physiopathology , Animals , Cell Differentiation , Epidermis/abnormalities , Epidermis/enzymology , Female , Hair Follicle/abnormalities , Hair Follicle/enzymology , Humans , Keratinocytes/enzymology , Keratinocytes/physiology , Male , Mice , Mice, Inbred C57BL , Pituitary Gland/abnormalities , Pituitary Gland/enzymology , Sebaceous Glands/abnormalities , Sebaceous Glands/enzymology , Skin/enzymology , Skin Abnormalities , Sphingolipids/metabolism
16.
Nature ; 477(7364): 289-94, 2011 Sep 14.
Article in English | MEDLINE | ID: mdl-21921910

ABSTRACT

We report genome sequences of 17 inbred strains of laboratory mice and identify almost ten times more variants than previously known. We use these genomes to explore the phylogenetic history of the laboratory mouse and to examine the functional consequences of allele-specific variation on transcript abundance, revealing that at least 12% of transcripts show a significant tissue-specific expression bias. By identifying candidate functional variants at 718 quantitative trait loci we show that the molecular nature of functional variants and their position relative to genes vary according to the effect size of the locus. These sequences provide a starting point for a new era in the functional analysis of a key model organism.


Subject(s)
Gene Expression Regulation/genetics , Genetic Variation/genetics , Genome/genetics , Mice, Inbred Strains/genetics , Mice/genetics , Phenotype , Alleles , Animals , Animals, Laboratory/genetics , Genomics , Mice/classification , Mice, Inbred C57BL/genetics , Phylogeny , Quantitative Trait Loci/genetics
17.
PLoS Genet ; 10(2): e1004167, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24586197

ABSTRACT

Retroviral insertional mutagenesis (RIM) is a powerful tool for cancer genomics that was combined in this study with deep sequencing (RIM/DS) to facilitate a comprehensive analysis of lymphoma progression. Transgenic mice expressing two potent collaborating oncogenes in the germ line (CD2-MYC, -Runx2) develop rapid onset tumours that can be accelerated and rendered polyclonal by neonatal Moloney murine leukaemia virus (MoMLV) infection. RIM/DS analysis of 28 polyclonal lymphomas identified 771 common insertion sites (CISs) defining a 'progression network' that encompassed a remarkably large fraction of known MoMLV target genes, with further strong indications of oncogenic selection above the background of MoMLV integration preference. Progression driven by RIM was characterised as a Darwinian process of clonal competition engaging proliferation control networks downstream of cytokine and T-cell receptor signalling. Enhancer mode activation accounted for the most efficiently selected CIS target genes, including Ccr7 as the most prominent of a set of chemokine receptors driving paracrine growth stimulation and lymphoma dissemination. Another large target gene subset including candidate tumour suppressors was disrupted by intragenic insertions. A second RIM/DS screen comparing lymphomas of wild-type and parental transgenics showed that CD2-MYC tumours are virtually dependent on activation of Runx family genes in strong preference to other potent Myc collaborating genes (Gfi1, Notch1). Ikzf1 was identified as a novel collaborating gene for Runx2 and illustrated the interface between integration preference and oncogenic selection. Lymphoma target genes for MoMLV can be classified into (a) a small set of master regulators that confer self-renewal; overcoming p53 and other failsafe pathways and (b) a large group of progression genes that control autonomous proliferation in transformed cells. These findings provide insights into retroviral biology, human cancer genetics and the safety of vector-mediated gene therapy.


Subject(s)
Genes, myb/genetics , Lymphoma/genetics , Moloney murine leukemia virus/genetics , Mutagenesis, Insertional/genetics , Tumor Suppressor Protein p53/genetics , Animals , Carcinogenesis , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Germ Cells , High-Throughput Nucleotide Sequencing , Humans , Ikaros Transcription Factor/biosynthesis , Ikaros Transcription Factor/genetics , Lymphoma/pathology , Lymphoma/virology , Mice
18.
EMBO J ; 31(11): 2486-97, 2012 May 30.
Article in English | MEDLINE | ID: mdl-22510880

ABSTRACT

Genetic screens in simple model organisms have identified many of the key components of the conserved signal transduction pathways that are oncogenic when misregulated. Here, we identify H37N21.1 as a gene that regulates vulval induction in let-60(n1046gf), a strain with a gain-of-function mutation in the Caenorhabditis elegans Ras orthologue, and show that somatic deletion of Nrbp1, the mouse orthologue of this gene, results in an intestinal progenitor cell phenotype that leads to profound changes in the proliferation and differentiation of all intestinal cell lineages. We show that Nrbp1 interacts with key components of the ubiquitination machinery and that loss of Nrbp1 in the intestine results in the accumulation of Sall4, a key mediator of stem cell fate, and of Tsc22d2. We also reveal that somatic loss of Nrbp1 results in tumourigenesis, with haematological and intestinal tumours predominating, and that nuclear receptor binding protein 1 (NRBP1) is downregulated in a range of human tumours, where low expression correlates with a poor prognosis. Thus NRBP1 is a conserved regulator of cell fate, that plays an important role in tumour suppression.


Subject(s)
Homeostasis/physiology , Intestines/physiology , Intracellular Signaling Peptides and Proteins/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Stem Cells/physiology , Tumor Suppressor Proteins/genetics , Vesicular Transport Proteins/physiology , Animals , Carrier Proteins/analysis , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , DNA-Binding Proteins/analysis , Female , Gene Deletion , Humans , Intestines/cytology , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Oxidoreductases , Prognosis , Receptors, Cytoplasmic and Nuclear/genetics , Stem Cells/cytology , Transcription Factors/analysis , Tumor Suppressor Proteins/physiology , Ubiquitination/genetics , Ubiquitination/physiology , Vesicular Transport Proteins/genetics
19.
Blood ; 124(24): 3624-35, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25258341

ABSTRACT

NBEAL2 encodes a multidomain scaffolding protein with a putative role in granule ontogeny in human platelets. Mutations in NBEAL2 underlie gray platelet syndrome (GPS), a rare inherited bleeding disorder characterized by a lack of α-granules within blood platelets and progressive bone marrow fibrosis. We present here a novel Nbeal2(-/-) murine model of GPS and demonstrate that the lack of α-granules is due to their loss from platelets/mature megakaryocytes (MKs), and not by initial impaired formation. We show that the lack of Nbeal2 confers a proinflammatory phenotype to the bone marrow MKs, which in combination with the loss of proteins from α-granules drives the development of bone marrow fibrosis. In addition, we demonstrate that α-granule deficiency impairs platelet function beyond their purely hemostatic role and that Nbeal2 deficiency has a protective effect against cancer metastasis.


Subject(s)
Gray Platelet Syndrome/metabolism , Megakaryocytes/metabolism , Animals , Blood Proteins/genetics , Blood Proteins/metabolism , Disease Models, Animal , Gray Platelet Syndrome/genetics , Gray Platelet Syndrome/pathology , Humans , Megakaryocytes/pathology , Mice , Mice, Knockout , Mutation , Neoplasm Metastasis , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Primary Myelofibrosis/genetics , Primary Myelofibrosis/metabolism , Primary Myelofibrosis/pathology , Secretory Vesicles
20.
BMC Cancer ; 15: 585, 2015 Aug 13.
Article in English | MEDLINE | ID: mdl-26269126

ABSTRACT

BACKGROUND: B-cell precursor acute lymphoblastic leukemia (B-ALL) is amongst the leading causes of childhood cancer-related mortality. Its most common chromosomal aberration is the ETV6-RUNX1 fusion gene, with ~25% of ETV6-RUNX1 patients also carrying PAX5 alterations. METHODS: We have recreated this mutation background by inter-crossing Etv6-RUNX1 (Etv6 (RUNX1-SB)) and Pax5(+/-) mice and performed an in vivo analysis to find driver genes using Sleeping Beauty transposon-mediated mutagenesis and also exome sequencing. RESULTS: Combination of Etv6-RUNX1 and Pax5(+/-) alleles generated a transplantable B220 + CD19+ B-ALL with a significant disease incidence. RNA-seq analysis showed a gene expression pattern consistent with arrest at the pre-B stage. Analysis of the transposon common insertion sites identified genes involved in B-cell development (Zfp423) and the JAK/STAT signaling pathway (Jak1, Stat5 and Il2rb), while exome sequencing revealed somatic hotspot mutations in Jak1 and Jak3 at residues analogous to those mutated in human leukemias, and also mutation of Trp53. CONCLUSIONS: Powerful synergies exists in our model suggesting STAT pathway activation and mutation of Trp53 are potent drivers of B-ALL in the context of Etv6-RUNX1;Pax5(+/-).


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Oncogene Proteins, Fusion/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins c-ets/genetics , Repressor Proteins/genetics , Animals , Disease Models, Animal , Janus Kinases/genetics , Mice , Mutagenesis , PAX5 Transcription Factor/genetics , ETS Translocation Variant 6 Protein
SELECTION OF CITATIONS
SEARCH DETAIL